Forkhead box protein O1

Last updated
FOXO1
Protein FOXO1 PDB 3CO6.png
Available structures
PDB Ortholog search: PDBe RCSB
Identifiers
Aliases FOXO1 , FKH1, FKHR, FOXO1A, forkhead box O1
External IDs OMIM: 136533 MGI: 1890077 HomoloGene: 1527 GeneCards: FOXO1
Orthologs
SpeciesHumanMouse
Entrez
Ensembl
UniProt
RefSeq (mRNA)

NM_002015

NM_019739

RefSeq (protein)

NP_002006

NP_062713

Location (UCSC) Chr 13: 40.56 – 40.67 Mb Chr 3: 52.18 – 52.26 Mb
PubMed search [3] [4]
Wikidata
View/Edit Human View/Edit Mouse

Forkhead box protein O1 (FOXO1), also known as forkhead in rhabdomyosarcoma (FKHR), is a protein that in humans is encoded by the FOXO1 gene. [5] FOXO1 is a transcription factor that plays important roles in regulation of gluconeogenesis and glycogenolysis by insulin signaling, and is also central to the decision for a preadipocyte to commit to adipogenesis. [6] It is primarily regulated through phosphorylation on multiple residues; its transcriptional activity is dependent on its phosphorylation state. [7] [8]

Contents

Function

Adipogenesis

FOXO1-dependent inhibition of adipogenesis FOXO1 adipogenesis.jpg
FOXO1-dependent inhibition of adipogenesis

FOXO1 negatively regulates adipogenesis. [9] Presently, the exact mechanism by which this is accomplished is not entirely understood. In the currently accepted model, FOXO1 negatively regulates adipogenesis by binding to the promoter sites of PPARG and preventing its transcription. Rising levels of PPARG are required to initiate adipogenesis; by preventing its transcription, FOXO1 is preventing the onset of adipogenesis. During stimulation by insulin, FOXO1 is excluded from the nucleus and is subsequently unable to prevent transcription of PPARG and inhibit adipogenesis. [10] However, there is substantial evidence to suggest that there are other factors that mediate the interaction between FOXO1 and the PPARG promoter, and that inhibition of adipogenesis is not entirely dependent on FOXO1 preventing transcription of PPARG. [11] The failure to commit to adipogenesis is primarily due to active FOXO1 arresting the cell in G0/G1 through activation of yet unknown downstream targets, with a putative target being SOD2. [12]

FOXO1 belongs to the forkhead family of transcription factors that are characterized by a distinct fork head domain. The specific function of this gene has not yet been determined; however, it may play a role in myogenic growth and differentiation. [13] FOXO1 is essential for the maintenance of human ESC pluripotency. This function is probably mediated through direct control by FOXO1 of OCT4 and SOX2 gene expression through occupation and activation of their respective promoters. [14] In hepatic cells this transcription factor seems to increase the expression of PEPCK and glycogen-6-phosphatase (the same enzymes that are blocked via the metformin/AMPK/SHP pathway). Blocking this transcription factor offers an opportunity for novel therapies for diabetes mellitus. [15] In pancreatic alpha-cells FOXO1 is important in regulating prepro-glucagon expression. [16] In pancreatic beta cells FOXO1 mediates glucagon-like peptide-1 effects on pancreatic beta-cell mass. [17]

Gluconeogenesis and glycogenolysis

Depicts insulin-regulated nuclear exclusion of FOXO1 and its effect on transcription of glucose-6 phosphatase Foxo1 glucose stim.pdf
Depicts insulin-regulated nuclear exclusion of FOXO1 and its effect on transcription of glucose-6 phosphatase

When the level of blood glucose is high, the pancreas releases insulin into the bloodstream. Insulin then causes the activation of PI3K, which subsequently phosphorylates Akt. Akt then phosphorylates FOXO1, causing nuclear exclusion. [18] [19] This phosphorylated FOXO1 is then ubiquitinated and degraded by the proteosome. [20] The phosphorylation of FOXO1 is irreversible; this prolongs insulin's inhibitory effect on glucose metabolism and hepatic glucose production. Transcription of glucose 6-phosphatase subsequently decreases, which consequently decreases the rates of gluconeogenesis and glycogenolysis. [21] FOXO1 also activates transcription of phosphoenolpyruvate carboxykinase, which is required for gluconeogenesis. [22] The activity of FOXO1 is also regulated through CBP induced acetylation [23] on Lys-242, Lys-245, and Lys-262. These lysine residues are located within the DNA-binding domain; acetylation inhibits the ability of FOXO1 to interact with the glucose-6 phosphatase promoter by decreasing the stability of the FOXO1-DNA complex. Additionally, this acetylation increases the rate of phosphorylation on Ser-253 by Akt. Mutating Ser-253 to Ala-253 makes FOXO1 constitutively active. SIRT1 reverses this acetylation process; however, the exact mechanism by which SIRT1 deacetylates FOXO1 is still under investigation; presently, acetylation is thought to mitigate the transcriptional activity of FOXO1 and thereby provide an additional level of metabolic regulation that is independent of the insulin/PI3K pathway. [24]

Apoptosis

FOXO1 may play an important role in apoptosis because it is phosphorylated and inhibited by AKT. [25] When FOXO1 is over expressed in human LNCaP prostate cancer cells, it causes apoptosis. [25] Also, FOXO1 regulates TNF-related apoptosis-inducing ligand (TRAIL), which caused FOXO1-induced apoptosis in the human prostate cancer cell line LAPC4 when FOXO1 adenovirus-mediated overexpression was used. [25] FOXO1 upregulates Fas ligand (FasL) transcriptionally resulting in apoptotic cell death. [25] Additionally, FOXO1 trans-activate Bim protein, which a member of the Bcl-2 family that promotes apoptosis and plays a role in the intrinsic mitochondrial apoptotic pathway. [25] Further, it was revealed that DNA damage-induced cell death in p53-deficient and p53-proficient cells was reduced when human FOXO1 is silenced by siRNA. [25] In type 2 diabetes the beta cells of the pancreas, which normally produce insulin undergo apoptosis, which greatly reduces insulin production. Fatty acids in the beta cells activate FOXO1, resulting in apoptosis of the beta cells. [26]


Cell Cycle Regulation

FOXO1 activation plays a role in cell cycle progression regulation. [25] The transcription and half- life of cyclin-dependent kinase inhibitor p27KIP1 rises when FOXO1 is active. [25] A study detects that FOXO1 regulates the nuclear localization of p27KIP1 in porcine granulosa cells and impacts cell cycle progression. [25] Furthermore, FOXO1-mediated cell cycle arrest is linked with cyclin D1 and cyclin D2 suppression in mammals. [25] It was detected that human FOXO1 is linked with the cyclin D1 promoter using chromatin immunoprecipitation assays (ChIP assays). [25] H215R is a human FOXO1 mutant, which cannot bind to the canonical FRE to induce expression of p27KIP1, repress cyclin D1 and cyclin D2 promoter activity and encourages cell cycle arrest at cyclin G1 (CCNG1). [25] As a result of that, activation of FOXO1 prevents the cell-division cycle at cyclin G1 (CCNG1) out of one of two ways stimulating or suppressing gene transcription. [25]

Mechanism of action

In its un-phosphorylated state, FOXO1 is localized to the nucleus, where it binds to the insulin response sequence located in the promoter for glucose 6-phosphatase and increases its rate of transcription. FOXO1, through increasing transcription of glucose-6-phosphatase, indirectly increases the rate of hepatic glucose production. [22] However, when FOXO1 is phosphorylated by Akt on Thr-24, Ser-256, and Ser-319, it is excluded from the nucleus, where it is then ubiquitinated and degraded. The phosphorylation of FOXO1 by Akt subsequently decreases the hepatic glucose production through a decrease in transcription of glucose 6-phosphatase.

Regulation

There are three processes, namely acetylation, phosphorylation, and ubiquitination that are responsible for regulation of the activity of forkhead box O1 (FOXO1). [27]

Phosphorylation

Phosphorylation of the FOXO1 protein is a result of the activation of the PI3K /AKT pathway. [27] Serum and glucocorticoid-inducible kinase SGK can also phosphorylate and inactivate FOXO1 transcription factor. [25] FOXO1 translocate from the nucleus to cytoplasm and inactivate through phosphorylation at well-defined sites by AKT/SGK1 protein kinases. [27] FOXO1 transcription factor can phosphorylate directly by AKT/SGK1 on three sites T24, S256 and S319. [28] Additionally, FOXO1 loses its interactions with DNA when phosphorylated by AKT/SGK1 because S256, which is one of the three AKT/SGK sites, changes the DNA-binding domain charge from a positive charge to a negative charge. [27]

Insulin signaling substrates 1 and 2 of the insulin-signaling cascade also regulate FOXO1 through phosphorylation by AKT. [27] AKT, which is referred to as protein kinase B, phosphorylates FOXO1 and accumulates in the cytosol. [27]

Casein kinase 1, a growth factor-activated protein kinase, also phosphorylates and potentiates FOXO1 and translocates FOXO1 to the cytoplasm. [27]

Research

Because FOXO1 provides a link between transcription and metabolic control by insulin, it is also a potential target for genetic control of type 2 diabetes. In the insulin-resistant murine model, there is increased hepatic glucose production due to a loss in insulin sensitivity; the rates of hepatic gluconeogenesis and glycogenolysis are increased when compared to normal mice; this is presumably due to un-regulated FOXO1. When the same experiment was repeated with haploinsufficient FOXO1, insulin sensitivity was partially restored, and hepatic glucose production subsequently decreased. [29] Similarly, in mice fed with a high fat diet (HFD), there is increased insulin resistance in skeletal and liver cells. However, when haploinsufficient FOXO1 mice were treated with the same HFD, there was a notable decrease in insulin resistance in both skeletal and liver cells. This effect was significantly augmented by the simultaneous administration of rosiglitazone, which is a commonly prescribed anti-diabetic drug. [30] These results create an opportunity for a novel gene therapy based approach to alleviating insulin desensitization in type 2 diabetes.

In diabetes (both type 1 and type 2), gluconeogenesis in the kidney contributes more to blood glucose than it does in normal subjects. [31] Enhancing suppression of FOXO1 by insulin can reduce gluconeogenesis in both the liver and kidney. [31]

In HFD-fed mice, the combination of FOXO1 and Notch-1 haploinsufficiency was more effective at restoring insulin sensitivity than FOXO1 haploinsufficiency alone. [32]

Insulin-producing cells could be generated through the inhibition of FOXO1 in intestinal organoids generated from intestinal stem cells isolated from adult tissue. [33]

Clinical significance

Interactions

FOXO1 has been shown to interact with:

Related Research Articles

<span class="mw-page-title-main">Cell cycle</span> Series of events and stages that result in cell division

The cell cycle, or cell-division cycle, is the series of events that take place in a cell that causes it to divide into two daughter cells. These events include the duplication of its DNA and some of its organelles, and subsequently the partitioning of its cytoplasm, chromosomes and other components into two daughter cells in a process called cell division.

<span class="mw-page-title-main">Pyruvate kinase</span> Class of enzymes

Pyruvate kinase is the enzyme involved in the last step of glycolysis. It catalyzes the transfer of a phosphate group from phosphoenolpyruvate (PEP) to adenosine diphosphate (ADP), yielding one molecule of pyruvate and one molecule of ATP. Pyruvate kinase was inappropriately named before it was recognized that it did not directly catalyze phosphorylation of pyruvate, which does not occur under physiological conditions. Pyruvate kinase is present in four distinct, tissue-specific isozymes in animals, each consisting of particular kinetic properties necessary to accommodate the variations in metabolic requirements of diverse tissues.

<span class="mw-page-title-main">Glucokinase</span> Enzyme participating to the regulation of carbohydrate metabolism

Glucokinase is an enzyme that facilitates phosphorylation of glucose to glucose-6-phosphate. Glucokinase occurs in cells in the liver and pancreas of humans and most other vertebrates. In each of these organs it plays an important role in the regulation of carbohydrate metabolism by acting as a glucose sensor, triggering shifts in metabolism or cell function in response to rising or falling levels of glucose, such as occur after a meal or when fasting. Mutations of the gene for this enzyme can cause unusual forms of diabetes or hypoglycemia.

<span class="mw-page-title-main">GSK-3</span> Class of enzymes

Glycogen synthase kinase 3 (GSK-3) is a serine/threonine protein kinase that mediates the addition of phosphate molecules onto serine and threonine amino acid residues. First discovered in 1980 as a regulatory kinase for its namesake, glycogen synthase (GS), GSK-3 has since been identified as a protein kinase for over 100 different proteins in a variety of different pathways. In mammals, including humans, GSK-3 exists in two isozymes encoded by two homologous genes GSK-3α (GSK3A) and GSK-3β (GSK3B). GSK-3 has been the subject of much research since it has been implicated in a number of diseases, including type 2 diabetes, Alzheimer's disease, inflammation, cancer, addiction and bipolar disorder.

<span class="mw-page-title-main">Restriction point</span> Animal cell cycle checkpoint

The restriction point (R), also known as the Start or G1/S checkpoint, is a cell cycle checkpoint in the G1 phase of the animal cell cycle at which the cell becomes "committed" to the cell cycle, and after which extracellular signals are no longer required to stimulate proliferation. The defining biochemical feature of the restriction point is the activation of G1/S- and S-phase cyclin-CDK complexes, which in turn phosphorylate proteins that initiate DNA replication, centrosome duplication, and other early cell cycle events. It is one of three main cell cycle checkpoints, the other two being the G2-M DNA damage checkpoint and the spindle checkpoint.

<span class="mw-page-title-main">Protein kinase B</span> Set of three serine/threonine-specific protein kinases

Protein kinase B (PKB), also known as Akt, is the collective name of a set of three serine/threonine-specific protein kinases that play key roles in multiple cellular processes such as glucose metabolism, apoptosis, cell proliferation, transcription, and cell migration.

In biochemistry, lipogenesis is the conversion of fatty acids and glycerol into fats, or a metabolic process through which acetyl-CoA is converted to triglyceride for storage in fat. Lipogenesis encompasses both fatty acid and triglyceride synthesis, with the latter being the process by which fatty acids are esterified to glycerol before being packaged into very-low-density lipoprotein (VLDL). Fatty acids are produced in the cytoplasm of cells by repeatedly adding two-carbon units to acetyl-CoA. Triacylglycerol synthesis, on the other hand, occurs in the endoplasmic reticulum membrane of cells by bonding three fatty acid molecules to a glycerol molecule. Both processes take place mainly in liver and adipose tissue. Nevertheless, it also occurs to some extent in other tissues such as the gut and kidney. A review on lipogenesis in the brain was published in 2008 by Lopez and Vidal-Puig. After being packaged into VLDL in the liver, the resulting lipoprotein is then secreted directly into the blood for delivery to peripheral tissues.

<span class="mw-page-title-main">Cyclin E</span> Member of the cyclin family

Cyclin E is a member of the cyclin family.

Nesfatin-1 is a neuropeptide produced in the hypothalamus of mammals. It participates in the regulation of hunger and fat storage. Increased nesfatin-1 in the hypothalamus contributes to diminished hunger, a 'sense of fullness', and a potential loss of body fat and weight.

The PHLPP isoforms are a pair of protein phosphatases, PHLPP1 and PHLPP2, that are important regulators of Akt serine-threonine kinases and conventional/novel protein kinase C (PKC) isoforms. PHLPP may act as a tumor suppressor in several types of cancer due to its ability to block growth factor-induced signaling in cancer cells.

<span class="mw-page-title-main">RELA</span> Protein-coding gene in the species Homo sapiens

Transcription factor p65 also known as nuclear factor NF-kappa-B p65 subunit is a protein that in humans is encoded by the RELA gene.

<span class="mw-page-title-main">AKT2</span> Protein-coding gene in the species Homo sapiens

AKT2, also known as RAC-beta serine/threonine-protein kinase, is an enzyme that in humans is encoded by the AKT2 gene. It influences metabolite storage as part of the insulin signal transduction pathway.

<span class="mw-page-title-main">FOXO3</span> Protein-coding gene in the species Homo sapiens

Forkhead box O3, also known as FOXO3 or FOXO3a, is a human protein encoded by the FOXO3 gene.

<span class="mw-page-title-main">FOXO4</span> Protein

Forkhead box protein O4 is a protein that in humans is encoded by the FOXO4 gene.

The Akt signaling pathway or PI3K-Akt signaling pathway is a signal transduction pathway that promotes survival and growth in response to extracellular signals. Key proteins involved are PI3K and Akt.

<span class="mw-page-title-main">Daf-16</span> Ortholog

DAF-16 is the sole ortholog of the FOXO family of transcription factors in the nematode Caenorhabditis elegans. It is responsible for activating genes involved in longevity, lipogenesis, heat shock survival and oxidative stress responses. It also protects C.elegans during food deprivation, causing it to transform into a hibernation - like state, known as a Dauer. DAF-16 is notable for being the primary transcription factor required for the profound lifespan extension observed upon mutation of the insulin-like receptor DAF-2. The gene has played a large role in research into longevity and the insulin signalling pathway as it is located in C. elegans, a successful ageing model organism.

<span class="mw-page-title-main">KLF15</span> Protein-coding gene in the species Homo sapiens

Krüppel-like factor 15 is a protein that in humans is encoded by the KLF15 gene in the Krüppel-like factor family. Its former designation KKLF stands for kidney-enriched Krüppel-like factor.

mTOR Complex 2 (mTORC2) is an acutely rapamycin-insensitive protein complex formed by serine/threonine kinase mTOR that regulates cell proliferation and survival, cell migration and cytoskeletal remodeling. The complex itself is rather large, consisting of seven protein subunits. The catalytic mTOR subunit, DEP domain containing mTOR-interacting protein (DEPTOR), mammalian lethal with sec-13 protein 8, and TTI1/TEL2 complex are shared by both mTORC2 and mTORC1. Rapamycin-insensitive companion of mTOR (RICTOR), mammalian stress-activated protein kinase interacting protein 1 (mSIN1), and protein observed with rictor 1 and 2 (Protor1/2) can only be found in mTORC2. Rictor has been shown to be the scaffold protein for substrate binding to mTORC2.

<span class="mw-page-title-main">FOXO6</span> Protein-coding gene in the species Homo sapiens

Forkhead box O6 is a protein that in humans is encoded by the FOXO6 gene.

Shaodong Guo is a Chinese-American nutrition scientist, academic, and diabetes researcher. He is a professor in nutrition and medicine at the Department of Nutrition of Texas A&M University. He has been a senior editor for Journal of Endocrinology and Journal of Molecular Endocrinology.

References

  1. 1 2 3 GRCh38: Ensembl release 89: ENSG00000150907 - Ensembl, May 2017
  2. 1 2 3 GRCm38: Ensembl release 89: ENSMUSG00000044167 - Ensembl, May 2017
  3. "Human PubMed Reference:". National Center for Biotechnology Information, U.S. National Library of Medicine.
  4. "Mouse PubMed Reference:". National Center for Biotechnology Information, U.S. National Library of Medicine.
  5. 1 2 Galili N, Davis RJ, Fredericks WJ, Mukhopadhyay S, Rauscher FJ, Emanuel BS, Rovera G, Barr FG (November 1993). "Fusion of a fork head domain gene to PAX3 in the solid tumour alveolar rhabdomyosarcoma". Nature Genetics. 5 (3): 230–5. doi:10.1038/ng1193-230. PMID   8275086. S2CID   12374322.
  6. Nakae J, Kitamura T, Kitamura Y, Biggs WH, Arden KC, Accili D (January 2003). "The forkhead transcription factor Foxo1 regulates adipocyte differentiation". Developmental Cell. 4 (1): 119–29. doi: 10.1016/S1534-5807(02)00401-X . PMID   12530968.
  7. Rena G, Guo S, Cichy SC, Unterman TG, Cohen P (June 1999). "Phosphorylation of the transcription factor forkhead family member FKHR by protein kinase B". The Journal of Biological Chemistry. 274 (24): 17179–83. doi: 10.1074/jbc.274.24.17179 . PMID   10358075.
  8. Guo S, Rena G, Cichy S, He X, Cohen P, Unterman T (June 1999). "Phosphorylation of serine 256 by protein kinase B disrupts transactivation by FKHR and mediates effects of insulin on insulin-like growth factor-binding protein-1 promoter activity through a conserved insulin response sequence". The Journal of Biological Chemistry. 274 (24): 17184–92. doi: 10.1074/jbc.274.24.17184 . PMID   10358076.
  9. Farmer SR (January 2003). "The forkhead transcription factor Foxo1: a possible link between obesity and insulin resistance". Molecular Cell. 11 (1): 6–8. doi: 10.1016/S1097-2765(03)00003-0 . PMID   12535515.
  10. Armoni M, Harel C, Karni S, Chen H, Bar-Yoseph F, Ver MR, Quon MJ, Karnieli E (July 2006). "FOXO1 represses peroxisome proliferator-activated receptor-gamma1 and -gamma2 gene promoters in primary adipocytes. A novel paradigm to increase insulin sensitivity". The Journal of Biological Chemistry. 281 (29): 19881–91. doi: 10.1074/jbc.M600320200 . PMID   16670091.
  11. Puigserver P, Rhee J, Donovan J, Walkey CJ, Yoon JC, Oriente F, Kitamura Y, Altomonte J, Dong H, Accili D, Spiegelman BM (May 2003). "Insulin-regulated hepatic gluconeogenesis through FOXO1-PGC-1alpha interaction". Nature. 423 (6939): 550–5. Bibcode:2003Natur.423..550P. doi: 10.1038/nature01667 . PMID   12754525.
  12. Adachi M, Osawa Y, Uchinami H, Kitamura T, Accili D, Brenner DA (April 2007). "The forkhead transcription factor FoxO1 regulates proliferation and transdifferentiation of hepatic stellate cells". Gastroenterology. 132 (4): 1434–46. doi: 10.1053/j.gastro.2007.01.033 . PMID   17408630.
  13. "Entrez Gene: FOXO1 forkhead box O1 (rhabdomyosarcoma)".
  14. Zhang X, Yalcin S, Lee DF, Yeh TY, Lee SM, Su J, Mungamuri SK, Rimmelé P, Kennedy M, Sellers R, Landthaler M, Tuschl T, Chi NW, Lemischka I, Keller G, Ghaffari S (July 2011). "FOXO1 is an essential regulator of pluripotency in human embryonic stem cells". Nature Cell Biology. 13 (9): 1092–9. doi:10.1038/ncb2293. PMC   4053529 . PMID   21804543.
  15. Nagashima T, Shigematsu N, Maruki R, Urano Y, Tanaka H, Shimaya A, Shimokawa T, Shibasaki M (November 2010). "Discovery of novel forkhead box O1 inhibitors for treating type 2 diabetes: improvement of fasting glycemia in diabetic db/db mice". Molecular Pharmacology. 78 (5): 961–70. doi:10.1124/mol.110.065714. PMID   20736318. S2CID   212576.
  16. McKinnon CM, Ravier MA, Rutter GA (December 2006). "FoxO1 is required for the regulation of preproglucagon gene expression by insulin in pancreatic alphaTC1-9 cells". The Journal of Biological Chemistry. 281 (51): 39358–69. doi: 10.1074/jbc.M605022200 . PMID   17062568.
  17. Buteau J, Spatz ML, Accili D (May 2006). "Transcription factor FoxO1 mediates glucagon-like peptide-1 effects on pancreatic beta-cell mass". Diabetes. 55 (5): 1190–6. doi: 10.2337/db05-0825 . PMID   16644672.
  18. Rena G, Woods YL, Prescott AR, Peggie M, Unterman TG, Williams MR, Cohen P (May 2002). "Two novel phosphorylation sites on FKHR that are critical for its nuclear exclusion". The EMBO Journal. 21 (9): 2263–71. doi:10.1093/emboj/21.9.2263. PMC   125977 . PMID   11980723.
  19. Rena G, Prescott AR, Guo S, Cohen P, Unterman TG (March 2001). "Roles of the forkhead in rhabdomyosarcoma (FKHR) phosphorylation sites in regulating 14-3-3 binding, transactivation and nuclear targetting". The Biochemical Journal. 354 (Pt 3): 605–12. doi:10.1042/0264-6021:3540605. PMC   1221692 . PMID   11237865.
  20. Matsuzaki H, Daitoku H, Hatta M, Tanaka K, Fukamizu A (September 2003). "Insulin-induced phosphorylation of FKHR (Foxo1) targets to proteasomal degradation". Proceedings of the National Academy of Sciences of the United States of America. 100 (20): 11285–90. Bibcode:2003PNAS..10011285M. doi: 10.1073/pnas.1934283100 . PMC   208749 . PMID   13679577.
  21. Daitoku H, Fukamizu A (June 2007). "FOXO transcription factors in the regulatory networks of longevity". Journal of Biochemistry. 141 (6): 769–74. doi:10.1093/jb/mvm104. PMID   17569704.
  22. 1 2 Nakae J, Kitamura T, Silver DL, Accili D (November 2001). "The forkhead transcription factor Foxo1 (Fkhr) confers insulin sensitivity onto glucose-6-phosphatase expression". The Journal of Clinical Investigation. 108 (9): 1359–67. doi:10.1172/JCI12876. PMC   209440 . PMID   11696581.
  23. Matsuzaki H, Daitoku H, Hatta M, Aoyama H, Yoshimochi K, Fukamizu A (August 2005). "Acetylation of Foxo1 alters its DNA-binding ability and sensitivity to phosphorylation". Proceedings of the National Academy of Sciences of the United States of America. 102 (32): 11278–83. Bibcode:2005PNAS..10211278M. doi: 10.1073/pnas.0502738102 . PMC   1183558 . PMID   16076959.
  24. Jing E, Gesta S, Kahn CR (August 2007). "SIRT2 regulates adipocyte differentiation through FoxO1 acetylation/deacetylation". Cell Metabolism. 6 (2): 105–14. doi:10.1016/j.cmet.2007.07.003. PMC   2083635 . PMID   17681146.
  25. 1 2 3 4 5 6 7 8 9 10 11 12 13 14 Lu H, Huang H (August 2011). "FOXO1: a potential target for human diseases". Current Drug Targets. 12 (9): 1235–44. doi:10.2174/138945011796150280. PMC   4591039 . PMID   21443466.
  26. Sun T, Han X (2019). "Death versus dedifferentiation: The molecular bases of beta cell mass reduction in type 2 diabetes". Seminars in Cell and Developmental Biology . 103: 76–82. doi:10.1016/j.semcdb.2019.12.002. PMID   31831356. S2CID   209341381.
  27. 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 Wang Y, Zhou Y, Graves DT (April 2014). "FOXO transcription factors: their clinical significance and regulation". BioMed Research International. 2014: 925350. doi: 10.1155/2014/925350 . PMC   4016844 . PMID   24864265.
  28. Hedrick SM, Hess Michelini R, Doedens AL, Goldrath AW, Stone EL (September 2012). "FOXO transcription factors throughout T cell biology". Nature Reviews. Immunology. 12 (9): 649–61. doi:10.1038/nri3278. PMC   3875397 . PMID   22918467.
  29. Nakae J, Biggs WH, Kitamura T, Cavenee WK, Wright CV, Arden KC, Accili D (October 2002). "Regulation of insulin action and pancreatic beta-cell function by mutated alleles of the gene encoding forkhead transcription factor Foxo1". Nature Genetics. 32 (2): 245–53. doi:10.1038/ng890. PMID   12219087. S2CID   12234973.
  30. Kim JJ, Li P, Huntley J, Chang JP, Arden KC, Olefsky JM (June 2009). "FoxO1 haploinsufficiency protects against high-fat diet-induced insulin resistance with enhanced peroxisome proliferator-activated receptor gamma activation in adipose tissue". Diabetes. 58 (6): 1275–82. doi:10.2337/db08-1001. PMC   2682681 . PMID   19289458.
  31. 1 2 Swe MT, Pongchaidecha A, Chatsudthipong V, Chattipakorn N, Lungkaphin A (2019). "Molecular signaling mechanisms of renal gluconeogenesis in nondiabetic and diabetic conditions". Journal of Cellular Physiology . 234 (6): 8134–8151. doi:10.1002/jcp.27598. PMID   30370538. S2CID   53097552.
  32. Pajvani UB, Shawber CJ, Samuel VT, Birkenfeld AL, Shulman GI, Kitajewski J, Accili D (July 2011). "Inhibition of Notch signaling ameliorates insulin resistance in a FoxO1-dependent manner". Nature Medicine. 17 (8): 961–7. doi:10.1038/nm.2378. PMC   3387563 . PMID   21804540.
  33. Bouchi R, Foo KS, Hua H, Tsuchiya K, Ohmura Y, Sandoval PR, Ratner LE, Egli D, Leibel RL, Accili D (June 2014). "FOXO1 inhibition yields functional insulin-producing cells in human gut organoid cultures". Nature Communications. 5: 4242. Bibcode:2014NatCo...5.4242B. doi:10.1038/ncomms5242. PMC   4083475 . PMID   24979718.
  34. Linardic CM (October 2008). "PAX3-FOXO1 fusion gene in rhabdomyosarcoma". Cancer Letters. 270 (1): 10–8. doi:10.1016/j.canlet.2008.03.035. PMC   2575376 . PMID   18457914.
  35. 1 2 3 Xiao E, Graves DT (August 2015). "Impact of Diabetes on the Protective Role of FOXO1 in Wound Healing". Journal of Dental Research. 94 (8): 1025–6. doi:10.1177/0022034515586353. PMC   530387 . PMID   25978971.
  36. Li P, Lee H, Guo S, Unterman TG, Jenster G, Bai W (January 2003). "AKT-independent protection of prostate cancer cells from apoptosis mediated through complex formation between the androgen receptor and FKHR". Molecular and Cellular Biology. 23 (1): 104–18. doi:10.1128/MCB.23.1.104-118.2003. PMC   140652 . PMID   12482965.
  37. Schuur ER, Loktev AV, Sharma M, Sun Z, Roth RA, Weigel RJ (September 2001). "Ligand-dependent interaction of estrogen receptor-alpha with members of the forkhead transcription factor family". The Journal of Biological Chemistry. 276 (36): 33554–60. doi: 10.1074/jbc.M105555200 . PMID   11435445.
  38. Nasrin N, Ogg S, Cahill CM, Biggs W, Nui S, Dore J, Calvo D, Shi Y, Ruvkun G, Alexander-Bridges MC (September 2000). "DAF-16 recruits the CREB-binding protein coactivator complex to the insulin-like growth factor binding protein 1 promoter in HepG2 cells". Proceedings of the National Academy of Sciences of the United States of America. 97 (19): 10412–7. Bibcode:2000PNAS...9710412N. doi: 10.1073/pnas.190326997 . PMC   27038 . PMID   10973497.
  39. Cao Y, Kamioka Y, Yokoi N, Kobayashi T, Hino O, Onodera M, Mochizuki N, Nakae J (December 2006). "Interaction of FoxO1 and TSC2 induces insulin resistance through activation of the mammalian target of rapamycin/p70 S6K pathway". The Journal of Biological Chemistry. 281 (52): 40242–51. doi: 10.1074/jbc.M608116200 . PMID   17077083.

This article incorporates text from the United States National Library of Medicine, which is in the public domain.